1
|
Singh AA, Yadav D, Khan F, Song M. Indole-3-Carbinol and Its Derivatives as Neuroprotective Modulators. Brain Sci 2024; 14:674. [PMID: 39061415 PMCID: PMC11274471 DOI: 10.3390/brainsci14070674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/26/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its downstream tropomyosin receptor kinase B (TrkB) signaling pathway play pivotal roles in the resilience and action of antidepressant drugs, making them prominent targets in psychiatric research. Oxidative stress (OS) contributes to various neurological disorders, including neurodegenerative diseases, stroke, and mental illnesses, and exacerbates the aging process. The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant responsive element (ARE) serves as the primary cellular defense mechanism against OS-induced brain damage. Thus, Nrf2 activation may confer endogenous neuroprotection against OS-related cellular damage; notably, the TrkB/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, stimulated by BDNF-dependent TrkB signaling, activates Nrf2 and promotes its nuclear translocation. However, insufficient neurotrophin support often leads to the downregulation of the TrkB signaling pathway in brain diseases. Thus, targeting TrkB activation and the Nrf2-ARE system is a promising therapeutic strategy for treating neurodegenerative diseases. Phytochemicals, including indole-3-carbinol (I3C) and its metabolite, diindolylmethane (DIM), exhibit neuroprotective effects through BDNF's mimetic activity; Akt phosphorylation is induced, and the antioxidant defense mechanism is activated by blocking the Nrf2-kelch-like ECH-associated protein 1 (Keap1) complex. This review emphasizes the therapeutic potential of I3C and its derivatives for concurrently activating neuronal defense mechanisms in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| | - Fazlurrahman Khan
- Institute of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea;
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (A.A.S.); (D.Y.)
| |
Collapse
|
2
|
Walzik D, Wences Chirino TY, Zimmer P, Joisten N. Molecular insights of exercise therapy in disease prevention and treatment. Signal Transduct Target Ther 2024; 9:138. [PMID: 38806473 PMCID: PMC11133400 DOI: 10.1038/s41392-024-01841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Despite substantial evidence emphasizing the pleiotropic benefits of exercise for the prevention and treatment of various diseases, the underlying biological mechanisms have not been fully elucidated. Several exercise benefits have been attributed to signaling molecules that are released in response to exercise by different tissues such as skeletal muscle, cardiac muscle, adipose, and liver tissue. These signaling molecules, which are collectively termed exerkines, form a heterogenous group of bioactive substances, mediating inter-organ crosstalk as well as structural and functional tissue adaption. Numerous scientific endeavors have focused on identifying and characterizing new biological mediators with such properties. Additionally, some investigations have focused on the molecular targets of exerkines and the cellular signaling cascades that trigger adaption processes. A detailed understanding of the tissue-specific downstream effects of exerkines is crucial to harness the health-related benefits mediated by exercise and improve targeted exercise programs in health and disease. Herein, we review the current in vivo evidence on exerkine-induced signal transduction across multiple target tissues and highlight the preventive and therapeutic value of exerkine signaling in various diseases. By emphasizing different aspects of exerkine research, we provide a comprehensive overview of (i) the molecular underpinnings of exerkine secretion, (ii) the receptor-dependent and receptor-independent signaling cascades mediating tissue adaption, and (iii) the clinical implications of these mechanisms in disease prevention and treatment.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Tiffany Y Wences Chirino
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, 37075, Göttingen, Lower Saxony, Germany.
| |
Collapse
|
3
|
Wang X, Yang C, Ma X, Li X, Qi Y, Bai Z, Xu Y, Ma K, Luo Y, Song J, Jia W, He Z, Liu Z. A division-of-labor mode contributes to the cardioprotective potential of mesenchymal stem/stromal cells in heart failure post myocardial infarction. Front Immunol 2024; 15:1363517. [PMID: 38562923 PMCID: PMC10982400 DOI: 10.3389/fimmu.2024.1363517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Background Treatment of heart failure post myocardial infarction (post-MI HF) with mesenchymal stem/stromal cells (MSCs) holds great promise. Nevertheless, 2-dimensional (2D) GMP-grade MSCs from different labs and donor sources have different therapeutic efficacy and still in a low yield. Therefore, it is crucial to increase the production and find novel ways to assess the therapeutic efficacy of MSCs. Materials and methods hUC-MSCs were cultured in 3-dimensional (3D) expansion system for obtaining enough cells for clinical use, named as 3D MSCs. A post-MI HF mouse model was employed to conduct in vivo and in vitro experiments. Single-cell and bulk RNA-seq analyses were performed on 3D MSCs. A total of 125 combination algorithms were leveraged to screen for core ligand genes. Shinyapp and shinycell workflows were used for deploying web-server. Result 3D GMP-grade MSCs can significantly and stably reduce the extent of post-MI HF. To understand the stable potential cardioprotective mechanism, scRNA-seq revealed the heterogeneity and division-of-labor mode of 3D MSCs at the cellular level. Specifically, scissor phenotypic analysis identified a reported wound-healing CD142+ MSCs subpopulation that is also associated with cardiac protection ability and CD142- MSCs that is in proliferative state, contributing to the cardioprotective function and self-renewal, respectively. Differential expression analysis was conducted on CD142+ MSCs and CD142- MSCs and the differentially expressed ligand-related model was achieved by employing 125 combination algorithms. The present study developed a machine learning predictive model based on 13 ligands. Further analysis using CellChat demonstrated that CD142+ MSCs have a stronger secretion capacity compared to CD142- MSCs and Flow cytometry sorting of the CD142+ MSCs and qRT-PCR validation confirmed the significant upregulation of these 13 ligand factors in CD142+ MSCs. Conclusion Clinical GMP-grade 3D MSCs could serve as a stable cardioprotective cell product. Using scissor analysis on scRNA-seq data, we have clarified the potential functional and proliferative subpopulation, which cooperatively contributed to self-renewal and functional maintenance for 3D MSCs, named as "division of labor" mode of MSCs. Moreover, a ligand model was robustly developed for predicting the secretory efficacy of MSCs. A user-friendly web-server and a predictive model were constructed and available (https://wangxc.shinyapps.io/3D_MSCs/).
Collapse
Affiliation(s)
- Xicheng Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Chao Yang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Xiaoxue Ma
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Xiuhua Li
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Yiyao Qi
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Zhihui Bai
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Ying Xu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Keming Ma
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Yi Luo
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Jiyang Song
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Zhiying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| | - Zhongmin Liu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| |
Collapse
|
4
|
Boukhatem I, Fleury S, Jourdi G, Lordkipanidzé M. The intriguing role of platelets as custodians of brain-derived neurotrophic factor. Res Pract Thromb Haemost 2024; 8:102398. [PMID: 38706782 PMCID: PMC11066552 DOI: 10.1016/j.rpth.2024.102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/26/2024] [Accepted: 03/18/2024] [Indexed: 05/07/2024] Open
Abstract
A State of the Art lecture titled "Platelets and neurotrophins" was presented at the International Society on Thrombosis and Haemostasis Congress in 2023. Neurotrophins, a family of neuronal growth factors known to support cognitive function, are increasingly recognized as important players in vascular health. Indeed, along with their canonical receptors, neurotrophins are expressed in peripheral tissues, particularly in the vasculature. The better-characterized neurotrophin in vascular biology is the brain-derived neurotrophic factor (BDNF). Its largest extracerebral pool resides within platelets, partly inherited from megakaryocytes and also likely internalized from circulation. Activation of platelets releases vast amounts of BDNF into their milieu and interestingly leads to platelet aggregation through binding of its receptor, the tropomyosin-related kinase B, on the platelet surface. As BDNF is readily available in plasma, a mechanism to preclude excessive platelet activation and aggregation appears critical. As such, binding of BDNF to α2-macroglobulin hinders its ability to bind its receptor and limits its platelet-activating effects to the site of vascular injury. Altogether, addition of BDNF to a forming clot facilitates not only paracrine platelet activation but also binding to fibrinogen, rendering the resulting clot more porous and plasma-permeable. Importantly, release of BDNF into circulation also appears to be protective against adverse cardiovascular and cerebrovascular outcomes, which has been reported in both animal models and epidemiologic studies. This opens an avenue for platelet-based strategies to deliver BDNF to vascular lesions and facilitate wound healing through its regenerative properties. Finally, we summarize relevant new data on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress.
Collapse
Affiliation(s)
- Imane Boukhatem
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Samuel Fleury
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
- Université Paris Cité, Institut National de la Santé Et de la Recherche Médicale, Innovative Therapies in Haemostasis, Paris, France
- Service d’Hématologie Biologique, Assistance Publique : Hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Amadio P, Porro B, Cavalca V, Zarà M, Eligini S, Sandrini L, Werba JP, Cosentino N, Olivares P, Galotta A, Bonomi A, Tremoli E, Trabattoni D, Barbieri SS. Hemostatic system in Takotsubo patients at long-term follow-up: A hidden activation? Int J Cardiol 2023; 390:131229. [PMID: 37527756 DOI: 10.1016/j.ijcard.2023.131229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Takotsubo cardiomyopathy (TTS) has long been considered a benign condition, despite recurrent events and long-term adverse outcomes are often reported. Endothelial damage, blood hyperviscosity, and platelet activation described in acute phase persist in long-term follow-up; however, TTS pathophysiology is still not fully understood. Here, we explored the hemostatic system at a median of 3.1 years after TTS to uncover additional long-lasting changes in these patients. METHODS We assessed hemostatic parameters in women with TTS (n = 23) or coronary artery disease (CAD; n = 31) and in control women (n = 26) age-matched, by thromboelastographic analysis, prothrombin time (PT) and partial thromboplastin time (aPTT) coagulation assays and microparticle exposing Tissue Factor (MP-TF). Functional fibrinogen and fibrin polymerization were analyzed by Clauss method and spectrophotometry, respectively. Platelet reactivity was evaluated by light transmission aggregometry, whereas plasminogen activator inhibitor-1 (PAI-1) and brain-derived neurotrophic factor (BDNF) were measured by ELISA kit. RESULTS Compared with control subjects, TTS patients exhibit an accelerated clot formation, higher percentage of fibrin polymerization and higher PAI-1 levels. Compared with CAD, TTS patients showed sustained residual platelet activation but decreased functional fibrinogen, fibrin polymerization and MP-TF levels, prolonged aPTT and a marked BDNF increase. CONCLUSIONS The long-term activation of hemostatic system observed in TTS patients compared to control subjects suggests a persistent humoral abnormality that may be related to the propensity for TTS recurrence. The higher residual platelet activity observed in TTS than in CAD patients invites investigation on TTS-tailored antiplatelet therapy potentially needed to prevent TTS adverse outcomes.
Collapse
Affiliation(s)
| | | | | | - Marta Zarà
- Centro Cardiologico Monzino I.R.C.C.S., Milan, Italy
| | - Sonia Eligini
- Centro Cardiologico Monzino I.R.C.C.S., Milan, Italy
| | | | | | | | | | | | - Alice Bonomi
- Centro Cardiologico Monzino I.R.C.C.S., Milan, Italy
| | - Elena Tremoli
- Maria Cecilia Hospital, GVM Care & Research, 48033, Cotignola, Ravenna, Italy
| | | | | |
Collapse
|
6
|
Larréché S, Chevillard L, Jourdi G, Mathé S, Servonnet A, Joly BS, Siguret V, Chippaux JP, Mégarbane B. Bothrops venom-induced hemostasis disorders in the rat: Between Scylla and Charybdis. PLoS Negl Trop Dis 2023; 17:e0011786. [PMID: 38011218 PMCID: PMC10703418 DOI: 10.1371/journal.pntd.0011786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/07/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
Hemostasis impairment represents the most threatening consequence of Viperidae envenoming, notably with Bothrops genus. In the French departments of America, B. atrox envenomation in French Guiana may lead to bleeding while B. lanceolatus envenomation in Martinique to thrombosis. Bleeding related to B. atrox envenomation is attributed to vascular damage mediated by venom metalloproteinases and blood uncoagulable state resulting from thrombocytopenia and consumptive coagulopathy. Thrombosis related to B. lanceolatus envenomation are poorly understood. We aimed to compare the effects of B. atrox and B. lanceolatus venoms in the rat to identify the determinants of the hemorrhagic versus thrombotic complications. Viscoelastometry (ROTEM), platelet count, plasma fibrinogen, thrombin generation assay, fibrinography, endothelial (von Willebrand factor, ADAMTS13 activity, ICAM-1, and soluble E-selectin), and inflammatory biomarkers (IL-1β, IL-6, TNF-α, MCP-1, and PAI-1) were determined in blood samples obtained at H3, H6, and H24 after the subcutaneous venom versus saline injection. In comparison to the control, initial fibrinogen consumption was observed with the two venoms while thrombocytopenia and reduction in the clot amplitude only with B. atrox venom. Moreover, we showed an increase in thrombin generation at H3 with the two venoms, an increase in fibrin generation accompanied with hyperfibrinogenemia at H24 and an increase in inflammatory biomarkers with B. lanceolatus venom. No endothelial damage was found with the two venoms. To conclude, our data support two-sided hemostasis complications in Bothrops envenoming with an initial risk of hemorrhage related to platelet consumption and hypocoagulability followed by an increased risk of thrombosis promoted by the activated inflammatory response and rapid-onset fibrinogen restoration.
Collapse
Affiliation(s)
- Sébastien Larréché
- Université Paris Cité, Inserm UMRS-1144, Paris, France
- Department of Medical Biology, Bégin Military Teaching Hospital, Saint-Mandé, France
| | | | - Georges Jourdi
- Université Paris Cité, Inserm UMRS-1140, Innovative Therapies in Hemostasis, Paris, France
- Department of Biological Hematology, Lariboisière Hospital, APHP, Paris, France
| | - Simon Mathé
- Université Paris Cité, Inserm UMRS-1144, Paris, France
| | - Aurélie Servonnet
- Unité analyses biologiques, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Bérangère S. Joly
- Université Paris Cité, Inserm UMRS-1140, Innovative Therapies in Hemostasis, Paris, France
- Université Paris Cité, EA3518, Institut de Recherche Saint-Louis, Paris, France
| | - Virginie Siguret
- Université Paris Cité, Inserm UMRS-1140, Innovative Therapies in Hemostasis, Paris, France
- Department of Biological Hematology, Lariboisière Hospital, APHP, Paris, France
| | - Jean-Philippe Chippaux
- Université Paris Cité, Research Institute for Development, Mother, and Child in Tropical Environment: Pathogens, Health system and Epidemiological transition, Paris, France
| | - Bruno Mégarbane
- Université Paris Cité, Inserm UMRS-1144, Paris, France
- Department of Medical and Toxicological Critical Care, Lariboisière Hospital, Federation of Toxicology, APHP, Paris, France
| |
Collapse
|
7
|
Jourdi G, Boukhatem I, Barcelona PF, Fleury S, Welman M, Saragovi HU, Pasquali S, Lordkipanidzé M. Alpha-2-macroglobulin prevents platelet aggregation induced by brain-derived neurotrophic factor. Biochem Pharmacol 2023; 215:115701. [PMID: 37487878 DOI: 10.1016/j.bcp.2023.115701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/07/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
The brain-derived neurotrophic factor (BDNF) has been recently shown to have activating effects in isolated platelets. However, BDNF circulates in plasma and a mechanism to preclude constant activation of platelets appears necessary. Hence, we investigated the mechanism regulating BDNF bioavailability in blood. Protein-protein interactions were predicted by molecular docking and validated through immunoprecipitation. Platelet aggregation was assessed using light transmission aggregometry with washed platelets in response to classical agonists or BDNF, in the absence or presence of alpha-2-macroglobulin (α2M), and in platelet-rich plasma. BDNF signaling was assessed with phospho-blots. As little as 25% autologous plasma was sufficient to completely abolish platelet aggregation in response to BDNF. Docking predicted two forms of BDNF binding to native or activated α2M, in parallel and perpendicular arrangements, and the model suggested that the BDNF-α2M complex cannot bind to the high-affinity BDNF receptor, tropomyosin receptor kinase B (TrkB). Experimentally, native and activated α2M formed stable complexes with BDNF preventing BDNF-induced TrkB activation and signal transduction. Both native and activated α2M inhibited BDNF induced-platelet aggregation in a concentration-dependent manner with comparable half-maximal inhibitory concentrations (IC50≈ 125-150 nM). Our study implicates α2M as a physiological regulator of BDNF bioavailability, and as an inhibitor of BDNF-induced platelet activation in blood.
Collapse
Affiliation(s)
- Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada; Université Paris Cité, INSERM, Innovative Therapies in Haemostasis, F-75006 Paris, France; Service d'Hématologie Biologique, AP-HP, Hôpital Lariboisière, F-75010 Paris, France
| | - Imane Boukhatem
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Pablo F Barcelona
- Departamento de Bioquímica Clínica, Centro de Investigaciones en Bioquímica Clínica e, Inmunología (CIBICI-CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - Samuel Fleury
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Melanie Welman
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada
| | - H Uri Saragovi
- Lady Davis Institute-Jewish General Hospital, Montreal, QC H3T 1E2, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3A 0G4, Canada
| | - Samuela Pasquali
- Université Paris Cité, CNRS UMR 8038, Laboratoire Cibles Thérapeutiques et Conception de Médicaments, F-75006 Paris, France; Université Paris Cité, CNRS UMR 8251, Laboratoire Biologie Fonctionnelle et Adaptative, F-75006 Paris, France
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada.
| |
Collapse
|
8
|
Preliminary results of the cross-sectional associations of sedentary behavior and physical activity with serum brain-derived neurotrophic factor in adults with coronary heart disease. Sci Rep 2022; 12:19685. [PMID: 36385629 PMCID: PMC9669050 DOI: 10.1038/s41598-022-23706-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022] Open
Abstract
This is the first study to analyze the association of accelerometer-measured patterns of habitual physical activity (PA) and sedentary behavior (SB) with serum BDNF in individuals with coronary heart disease. A total of 30 individuals (M = 69.5 years; 80% men) participated in this pre-post study that aimed to test a multi-behavioral intervention. All participants underwent standardized measurement of anthropometric variables, blood collection, self-administered survey, and accelerometer-based measurement of PA and SB over seven days. Serum BDNF concentrations were measured using enzyme-linked immunosorbent assay kit. We applied separate multiple linear regression analysis to estimate the associations of baseline SB pattern measures, light and moderate-to-vigorous PA with serum BDNF (n = 29). Participants spent 508.7 ± 76.5 min/d in SB, 258.5 ± 71.2 min/d in light PA, and 21.2 ± 15.2 min/d in moderate-to-vigorous PA. Per day, individuals had 15.5 ± 3.2 numbers of 10-to-30 min bouts of SB (average length: 22.2 ± 2.1 min) and 3.4 ± 1.2 numbers of > 30 min bouts of SB (average length: 43.8 ± 2.4 min). Regression analysis revealed no significant associations between any of the accelerometer-based measures and serum BDNF. The findings of this study did not reveal an association of accelerometer-measured PA and SB pattern variables with serum BDNF in individuals with coronary heart disease. In addition, our data revealed a considerable variation of PA and SB which should be considered in future studies.
Collapse
|
9
|
Azoulay D, Naamad M, Frydman D, Broide E, Zimran A, Stemer G, Revel-Vilk S. Brain-Derived Neurotrophic Factor (BDNF) Is Associated with Platelet Activity and Bleeding Tendency in Patients with Gaucher Disease. Int J Mol Sci 2022; 23:13982. [PMID: 36430458 PMCID: PMC9697957 DOI: 10.3390/ijms232213982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Bleeding tendency, a prominent feature of patients with Gaucher disease (GD), is associated with abnormal platelet function. Brain-derived neurotrophic factor (BDNF) is a protein with neuroprotective potential stored in alpha granules of circulating platelets. Here we studied BDNF levels in 50 patients with type I GD (GD1) and their correlation with platelet activity and bleeding tendency. Flow cytometry was used to test unstimulated and stimulated measurement of platelet surface-activated expression of αIIbβ3 integrin, P-selectin and lysosomal-associated membrane protein (LAMP3/CD63). Serum and plasma BDNF levels were quantified using ELISA. The bleeding history was recorded by a bleeding questionnaire. Serum BDNF levels were positively correlated with platelet count and moderately correlated with unstimulated and stimulated platelet P-selectin expression. Patients with more than one bleeding manifestation were shown to have lower serum BDNF levels, albeit similar platelet count. Plasma BDNF levels were significantly elevated in splenectomized patients and showed a moderate positive correlation with stimulated platelet CD63 expression. These observations demonstrate the first association between BDNF levels in the peripheral blood with platelet dysfunction and increased bleeding manifestation. The role of measuring serum BDNF for assessing platelet alpha degranulation defects and bleeding risk in patients with GD and the general population needs further study.
Collapse
Affiliation(s)
- David Azoulay
- Hematology Unit and Laboratories, Galilee Medical Center, Nahariya 22100, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Mira Naamad
- Flow Cytometry Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Dafna Frydman
- Gaucher Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Ellen Broide
- Flow Cytometry Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Ari Zimran
- Gaucher Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Galia Stemer
- Hematology Unit and Laboratories, Galilee Medical Center, Nahariya 22100, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Shoshana Revel-Vilk
- Gaucher Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112002, Israel
- Pediatric Hematology/Oncology Unit, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| |
Collapse
|
10
|
The multifaceted role of platelets in mediating brain function. Blood 2022; 140:815-827. [PMID: 35609283 PMCID: PMC9412009 DOI: 10.1182/blood.2022015970] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Platelets, the small, anucleate blood cells that originate from megakaryocytes in the bone marrow, are typically associated with coagulation. However, it is now apparent that platelets are more multifaceted than originally thought, with their function extending beyond their traditional role in hemostasis to acting as important mediators of brain function. In this review, we outline the broad repertoire of platelet function in the central nervous system, focusing on the similarities between platelets and neurons. We also summarize the role that platelets play in the pathophysiology of various neurological diseases, with a particular focus on neuroinflammation and neurodegeneration. Finally, we highlight the exciting prospect of harnessing the unique features of the platelet proteome and extracellular vesicles, which are rich in neurotrophic, antioxidative, and antiinflammatory factors, for the development of novel neuroprotective and neuroregenerative interventions to treat various neurodegenerative and traumatic pathologies.
Collapse
|
11
|
Amadio P, Macchi C, Favero C, Zarà M, Solazzo G, Dioni L, Sandrini L, Vigna L, Greco MF, Buoli M, Sirtori CR, Pesatori AC, Ieraci A, Ruscica M, Barbieri SS, Bollati V. Brain-Derived Neurotrophic Factor and Extracellular Vesicle-Derived miRNAs in an Italian Cohort of Individuals With Obesity: A Key to Explain the Link Between Depression and Atherothrombosis. Front Cardiovasc Med 2022; 9:906483. [PMID: 35911513 PMCID: PMC9326054 DOI: 10.3389/fcvm.2022.906483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/16/2022] [Indexed: 12/28/2022] Open
Abstract
BackgroundObesity and depression are intertwined diseases often associated with an increased risk of cardiovascular (CV) complications. Brain-Derived Neurotrophic Factor (BDNF), altered in the brain both of subjects with depression and obesity, provides a potential link between depression and thrombosis. Since the relationship among peripheral BDNF, depression and obesity is not well-defined, the aim of the present report has been to address this issue taking advantage of the contribution played by extracellular vesicle (EV)-derived miRNAs.Research ProcessAssociations among circulating BDNF, depression and EV-derived miRNAs related to atherothrombosis have been evaluated in a large Italian cohort of obese individuals (n = 743), characterized by the Beck Depression Inventory (BDI-II) score.ResultsBDI-II was negatively associated with BDNF levels without a significant impact of the rs6265 BDNF polymorphism; this association was modified by raised levels of IFN-γ. BDNF levels were linked to an increase of 80 EV-derived miRNAs and a decrease of 59 miRNAs related to atherosclerosis and thrombosis. Network analysis identified at least 18 genes targeted by these miRNAs, 7 of which involved in depression and CV risk. The observation of a possible link among BDNF, depression, and miRNAs related to atherothrombosis and depression in obesity is novel and may lead to a wider use of BDNF as a CV risk biomarker in this specific subject group.
Collapse
Affiliation(s)
- Patrizia Amadio
- Brain-Heart Axis: Cellular and Molecular Mechanisms Unit, Centro Cardiologico Monzino Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Chiara Macchi
- Department of Biomolecular and Pharmacological Sciences, University of Milan, Milan, Italy
| | - Chiara Favero
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Marta Zarà
- Brain-Heart Axis: Cellular and Molecular Mechanisms Unit, Centro Cardiologico Monzino Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giulia Solazzo
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Laura Dioni
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Leonardo Sandrini
- Brain-Heart Axis: Cellular and Molecular Mechanisms Unit, Centro Cardiologico Monzino Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Luisella Vigna
- Occupational Health Unit, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Francesca Greco
- Department of Biomolecular and Pharmacological Sciences, University of Milan, Milan, Italy
| | - Massimiliano Buoli
- Department of Neurosciences and Mental Health, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca'Granda Ospedale, Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Cesare R. Sirtori
- Department of Biomolecular and Pharmacological Sciences, University of Milan, Milan, Italy
| | - Angela Cecilia Pesatori
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Occupational Health Unit, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandro Ieraci
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Massimiliano Ruscica
- Department of Biomolecular and Pharmacological Sciences, University of Milan, Milan, Italy
- *Correspondence: Massimiliano Ruscica
| | - Silvia Stella Barbieri
- Brain-Heart Axis: Cellular and Molecular Mechanisms Unit, Centro Cardiologico Monzino Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
- Silvia Stella Barbieri
| | - Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
12
|
Azoulay D, Horowitz NA. Brain-derived neurotrophic factor in hematological malignancies: From detrimental to potentially beneficial. Blood Rev 2021; 51:100871. [PMID: 34344590 DOI: 10.1016/j.blre.2021.100871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/13/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022]
Abstract
Emerging studies have highlighted brain-derived neurotrophic factor (BDNF), a neuronal growth factor abundant in the peripheral blood, and its tyrosine kinase receptor TRKB, as onco-genes and proteins that support the survival of malignant hematological cells. In contrast, other researchers reported on a favorable association between BDNF blood levels and prognosis, chemotherapy response and neurological side effects in patients with hematological malignancies. Here, we review the accumulated data regarding the expression of BDNF and its receptors in normal hematopoietic and lymphatic cells and tissue. In addition, in-vitro experiments, animal models and human sample studies that investigated the role of BDNF and its receptors in hematological malignancies are discussed. Finally, directions for future research aimed at revealing the mechanisms underlying the protective effect of BDNF in patients with these diseases are suggested.
Collapse
Affiliation(s)
- David Azoulay
- Hematology Unit and Laboratories, Galilee Medical Center, Naharia, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | - Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
13
|
Amadio P, Cosentino N, Eligini S, Barbieri S, Tedesco CC, Sandrini L, Zarà M, Fabiocchi F, Niccoli G, Magnani G, Fracassi F, Crea F, Veglia F, Marenzi G, Barbieri SS. Potential Relation between Plasma BDNF Levels and Human Coronary Plaque Morphology. Diagnostics (Basel) 2021; 11:diagnostics11061010. [PMID: 34205863 PMCID: PMC8226920 DOI: 10.3390/diagnostics11061010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/30/2021] [Indexed: 01/13/2023] Open
Abstract
Coronary artery disease (CAD) patients are at high ischemic risk, and new biomarkers reflecting atherosclerotic disease severity and coronary plaque vulnerability are required. The Brain-Derived Neurotrophic Factor (BDNF) affects endothelial and macrophage activation suggesting its involvement in atherosclerotic plaque behavior. To investigate whether plasma BDNF is associated with in vivo coronary plaque features, assessed by optical coherence tomography (OCT), in both acute myocardial infarction (AMI) and stable angina (SA) patients, we enrolled 55 CAD patients (31 SA and 24 AMI), and 21 healthy subjects (HS). BDNF was lower in CAD patients than in HS (p < 0.0001), and it decreased with the presence, clinical acuity and severity of CAD. The greater BDNF levels were associated with OCT features of plaque vulnerability in overall CAD as well as in SA and AMI patients (p < 0.03). Specifically, in SA patients, BDNF correlated positively with macrophages’ infiltration within atherosclerotic plaque (p = 0.01) and inversely with minimal lumen area (p = 0.02). In AMI patients a negative correlation between BDNF and cap thickness was found (p = 0.02). Despite a small study population, our data suggest a relationship between BDNF and coronary plaque vulnerability, showing that vulnerable plaque is positively associated with plasma BDNF levels, regardless of the clinical CAD manifestation.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
| | - Nicola Cosentino
- Intensive Cardiac Care Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (N.C.); (G.M.)
| | - Sonia Eligini
- Unit of Metabolomics and Cellular Biochemistry of Atherothrombosis, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy;
| | - Simone Barbieri
- Unit of Biostatistics, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (S.B.); (C.C.T.); (F.V.)
| | - Calogero Claudio Tedesco
- Unit of Biostatistics, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (S.B.); (C.C.T.); (F.V.)
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
| | - Franco Fabiocchi
- Interventional Cardiology Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy;
| | - Giampaolo Niccoli
- Cardiology Unit, Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (G.N.); (G.M.)
| | - Giulia Magnani
- Cardiology Unit, Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (G.N.); (G.M.)
| | - Francesco Fracassi
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (F.F.); (F.C.)
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (F.F.); (F.C.)
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (S.B.); (C.C.T.); (F.V.)
| | - Giancarlo Marenzi
- Intensive Cardiac Care Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (N.C.); (G.M.)
| | - Silvia Stella Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
- Correspondence: ; Tel.: +39-02-58002021
| |
Collapse
|
14
|
Circulating neurotrophins and hemostatic risk factors of atherothrombotic cardiovascular disease at baseline and during sympathetic challenge: the SABPA study. Sci Rep 2021; 11:2297. [PMID: 33504912 PMCID: PMC7841151 DOI: 10.1038/s41598-021-81946-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 01/14/2021] [Indexed: 01/15/2023] Open
Abstract
Sympathetic activation may trigger acute coronary syndromes. We examined the relation between circulating neurotrophic factors and hemostatic risk factors of atherothrombotic cardiovascular disease at baseline and in response to acute mental stress to establish a brain-heart link. In 409 black and white South Africans, brain-derived neurotrophic factor (BDNF) and fibrinolytic measures were assessed at baseline. Glial cell-derived neurotrophic factor (GDNF), S100 calcium-binding protein (S100B), von Willebrand factor (VWF), fibrinogen and D-dimer were assessed at baseline and 10 min after the Stroop test. Neurotrophins were regressed on hemostatic measures adjusting for demographics, comorbidities, cardiometabolic factors and health behaviors. Higher baseline BDNF was associated with greater stress-induced increase in fibrinogen (p = 0.003) and lower D-dimer increase (p = 0.016). Higher baseline S100B was significantly associated with higher baseline VWF (p = 0.031) and lower fibrinogen increase (p = 0.048). Lower baseline GDNF was associated with higher baseline VWF (p = 0.035) but lower VWF increase (p = 0.001). Greater GDNF (p = 0.006) and S100B (p = 0.042) increases were associated with lower VWF increase. All associations showed small-to-moderate effect sizes. Neurotrophins and fibrinolytic factors showed no significant associations. The findings support the existence of a peripheral neurothrophin-hemostasis interaction of small-to-moderate clinical relevance. The implications for atherothrombotic cardiovascular disease need further exploration.
Collapse
|
15
|
Evaluation of serum platelet-derived growth factor receptor-ß and brain-derived neurotrophic factor levels in microvascular angina. Anatol J Cardiol 2020; 24:397-404. [PMID: 33253128 PMCID: PMC7791298 DOI: 10.14744/anatoljcardiol.2020.44388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective: Microvascular angina (MVA) is a coronary microcirculation disease. Research on microcirculatory dysfunction has revealed several biomarkers involved in the etiopathogenesis of MVA. Platelet-derived growth factor receptor β (PDGFR-β) and brain-derived neurotrophic factor (BDNF) are 2 biomarkers associated with microcirculation, particularly pericytes function. The aim of this study was to investigate the role of PDGFR-β and BDNF in MVA. Methods: Ninety-one patients (median age, 56 y; age range, 40–79 y; 36 men) with MVA and 61 control group subjects (median age, 52 y; age range, 38–76 y; 29 men) were included in the study. Serum concentrations of PDGFR-β and BDNF were measured with commercially available enzyme-linked immunosorbent assay kits. Results: PDGFR-β [2.82 ng/ml; interquartile range (IQR), 0.57–7.79 ng/ml vs. 2.27 ng/ml; IQR, 0.41–7.16 ng/ml; p<0.0005] and BDNF (2.41 ng/ml; IQR, 0.97–7.97 ng/ml vs. 1.92 ng/ml; IQR, 1.07–6.67 ng/ml; p=0.023) concentrations were significantly higher in patients with MVA compared with the controls. PDGFR-β correlated positively with age (r=0.26, p=0.001), low-density lipoprotein (r=0.18; p=0.02), and BDNF (r=0.47; p<0.001), and BDNF showed a significant positive correlation with age (r=0.20; p=0.01). In binary logistic regression analysis, high-sensitivity C-reactive protein, uric acid, and PDGFR-β values were found to be independent predictors of MVA. Conclusion: MVA is associated with higher PDGFR-β and BDNF levels. This association may indicate an abnormality in microvascular function. Future studies are required to determine the role of these biomarkers in the pathogenesis of MVA. (Anatol J Cardiol 2020; 24: 397-404)
Collapse
|
16
|
Beyond Haemostasis and Thrombosis: Platelets in Depression and Its Co-Morbidities. Int J Mol Sci 2020; 21:ijms21228817. [PMID: 33233416 PMCID: PMC7700239 DOI: 10.3390/ijms21228817] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Alongside their function in primary haemostasis and thrombo-inflammation, platelets are increasingly considered a bridge between mental, immunological and coagulation-related disorders. This review focuses on the link between platelets and the pathophysiology of major depressive disorder (MDD) and its most frequent comorbidities. Platelet- and neuron-shared proteins involved in MDD are functionally described. Platelet-related studies performed in the context of MDD, cardiovascular disease, and major neurodegenerative, neuropsychiatric and neurodevelopmental disorders are transversally presented from an epidemiological, genetic and functional point of view. To provide a complete scenario, we report the analysis of original data on the epidemiological link between platelets and depression symptoms suggesting moderating and interactive effects of sex on this association. Epidemiological and genetic studies discussed suggest that blood platelets might also be relevant biomarkers of MDD prediction and occurrence in the context of MDD comorbidities. Finally, this review has the ambition to formulate some directives and perspectives for future research on this topic.
Collapse
|
17
|
Amadio P, Zarà M, Sandrini L, Ieraci A, Barbieri SS. Depression and Cardiovascular Disease: The Viewpoint of Platelets. Int J Mol Sci 2020; 21:E7560. [PMID: 33066277 PMCID: PMC7589256 DOI: 10.3390/ijms21207560] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Depression is a major cause of morbidity and low quality of life among patients with cardiovascular disease (CVD), and it is now considered as an independent risk factor for major adverse cardiovascular events. Increasing evidence indicates not only that depression worsens the prognosis of cardiac events, but also that a cross-vulnerability between the two conditions occurs. Among the several mechanisms proposed to explain this interplay, platelet activation is the more attractive, seeing platelets as potential mirror of the brain function. In this review, we dissected the mechanisms linking depression and CVD highlighting the critical role of platelet behavior during depression as trigger of cardiovascular complication. In particular, we will discuss the relationship between depression and molecules involved in the CVD (e.g., catecholamines, adipokines, lipids, reactive oxygen species, and chemokines), emphasizing their impact on platelet activation and related mechanisms.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| | - Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy;
| | - Silvia Stella Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanism, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (M.Z.); (L.S.)
| |
Collapse
|
18
|
Wu HB, Shao K, Wang YC, Wang XC, Liu HL, Xie YT, Du RP. Research progress of CA125 and BDNF in serum of patients with acute myocardial infarction for predicting acute heart failure. Clin Hemorheol Microcirc 2020; 75:99-106. [PMID: 31868662 DOI: 10.3233/ch-190738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Hai-Bo Wu
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Kai Shao
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yun-Can Wang
- Department of Ultrasound, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xue-Chao Wang
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Hui-Liang Liu
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Yue-Tao Xie
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Rong-Pin Du
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Vandaele J, Louis B, Liu K, Camacho R, Kouwer PHJ, Rocha S. Structural characterization of fibrous synthetic hydrogels using fluorescence microscopy. SOFT MATTER 2020; 16:4210-4219. [PMID: 32292943 DOI: 10.1039/c9sm01828j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The structural features of the matrix surrounding the cells play a crucial role in regulating their behavior. Here, we used fluorescence microscopy and customized analysis algorithms to characterize the architecture of fibrous hydrogel networks. As a model system, we investigated a new class of synthetic biomimetic material, hydrogels prepared from polyisocyanides. Our results show that these synthetic gels present a highly heterogeneous fibrous network, with pores reaching a few micrometers in diameter. By encapsulating HeLa cells in different hydrogels, we show that a more porous structure is linked to a higher proliferation rate. The approach described here, for the characterization of the network of fibrous hydrogels, can be easily applied to other polymer-based materials and provide new insights into the influence of structural features in cell behavior. This knowledge is crucial to develop the next generation of biomimetic materials for 3D cell models and tissue engineering applications.
Collapse
|
20
|
Impact of BDNF Val66Met Polymorphism on Myocardial Infarction: Exploring the Macrophage Phenotype. Cells 2020; 9:cells9051084. [PMID: 32349267 PMCID: PMC7290372 DOI: 10.3390/cells9051084] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin growth factor family, well known for its role in the homeostasis of the cardiovascular system. Recently, the human BDNF Val66Met single nucleotide polymorphism has been associated with the increased propensity for arterial thrombosis related to acute myocardial infarction (AMI). Using cardiac magnetic resonance imaging and immunohistochemistry analyses, we showed that homozygous mice carrying the human BDNF Val66Met polymorphism (BDNFMet/Met) undergoing left anterior descending (LAD) coronary artery ligation display an adverse cardiac remodeling compared to wild-type (BDNFVal/Val). Interestingly, we observed a persistent presence of pro-inflammatory M1-like macrophages and a reduced accumulation of reparative-like phenotype macrophages (M2-like) in the infarcted heart of mutant mice. Further qPCR analyses showed that BDNFMet/Met peritoneal macrophages are more pro-inflammatory and have a higher migratory ability compared to BDNFVal/Val ones. Finally, macrophages differentiated from circulating monocytes isolated from BDNFMet/Met patients with coronary heart disease displayed the same pro-inflammatory characteristics of the murine ones. In conclusion, the BDNF Val66Met polymorphism predisposes to adverse cardiac remodeling after myocardial infarction in a mouse model and affects macrophage phenotype in both humans and mice. These results provide a new cellular mechanism by which this human BDNF genetic variant could influence cardiovascular disease.
Collapse
|
21
|
Canobbio I. Blood platelets: Circulating mirrors of neurons? Res Pract Thromb Haemost 2019; 3:564-565. [PMID: 31624775 PMCID: PMC6781913 DOI: 10.1002/rth2.12254] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/13/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Ilaria Canobbio
- Department of Biology and BiotechnologyUniversity of PaviaPaviaItaly
| |
Collapse
|